Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Chemistry ; 23(22): 5188-5192, 2017 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-28207175

RESUMEN

Allosteric compounds that stimulate Hsp90 adenosine triphosphatase (ATPase) activity were rationally designed, showing anticancer potencies in the low micromolar to nanomolar range. In parallel, the mode of action of these compounds was clarified and a quantitative model that links the dynamic ligand-protein cross-talk to observed cellular and in vitro activities was developed. The results support the potential of using dynamics-based approaches to develop original mechanism-based cancer therapeutics.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Antineoplásicos/farmacología , Proteínas HSP90 de Choque Térmico/metabolismo , Adenosina Trifosfatasas/química , Regulación Alostérica , Antineoplásicos/química , Diseño de Fármacos , Proteínas HSP90 de Choque Térmico/química , Ligandos , Unión Proteica
2.
Cell Mol Life Sci ; 73(13): 2531-42, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26970978

RESUMEN

Metastatic prostate cancer is a lethal disease that remains incurable despite the recent approval of new drugs, thus making the development of alternative treatment approaches urgently needed. A more precise understanding of the molecular mechanisms underlying prostate cancer dissemination could lead to the identification of novel therapeutic targets for the design of efficient anti-metastatic strategies. MicroRNA (miRNAs) are endogenous, small non-coding RNA molecules acting as key regulators of gene expression at post-transcriptional level. It has been clearly established that altered miRNA expression is a common hallmark of cancer. In addition, emerging evidence suggests their direct involvement in the metastatic cascade. In this review, we present a comprehensive overview of the data generated in experimental tumor models indicating that specific miRNAs may impinge on the different stages of prostate cancer metastasis, including (i) the regulation of epithelial-to-mesenchymal transition and cell migration/invasion, (ii) the interplay between cancer cells and the surrounding stroma, (iii) the control of angiogenesis, (iv) the regulation of anoikis, and (v) the colonization of distant organs. Moreover, we show preliminary evidence of the clinical relevance of some of these miRNAs, in terms of association with tumor aggressiveness/dissemination and clinical outcome, as emerged from translation studies carried out in prostate cancer patient cohorts. We also discuss the potential and the current limitations of manipulating metastasis-related miRNAs, by mimicking or inhibiting them, as a strategy for the development of novel therapeutic approaches for the advanced disease.


Asunto(s)
MicroARNs/genética , Metástasis de la Neoplasia/genética , Próstata/patología , Neoplasias de la Próstata/genética , Animales , Movimiento Celular , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , MicroARNs/análisis , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/terapia , Próstata/irrigación sanguínea , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia
3.
J Transl Med ; 14: 25, 2016 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-26810896

RESUMEN

BACKGROUND: Diffuse malignant peritoneal mesothelioma (DMPM) is a rare and locally aggressive disease. DMPM prognosis is dismal, mainly due to the lack of effective treatment options and the development of new therapeutic strategies is urgently needed. In this context, novel immunotherapy approaches can be explored in an attempt to improve DMPM patients' survival. METHODS: We tested the efficacy of CpG-oligodeoxynucleotides (CpG-ODN), synthetic DNA sequences recognized by Toll-like receptor 9 and able to induce innate/adaptive immune response, in two DMPM orthotopic xenografts (MesoII and STO), which properly recapitulate the dissemination pattern of the disease in the peritoneal cavity. Severe combined immunodeficiency mice carrying DMPM xenografts were treated at different stages of tumor development with i.p. delivered CpG-ODN1826 for 4 weeks. CpG-ODN1826-induced modulation in the composition of peritoneal immune infiltrate was assessed by flow cytometry. RESULTS: When administered to early-stage tumors (i.e., 4 days after i.p. DMPM cell injection in mice), the agent exhibited impressive efficacy against MesoII by completely inhibiting tumor take and ascites development (no evidence of tumor masses and ascites in 6/6 mice at necropsy), and also impaired STO tumor take and growth (4/6 tumor-free mice; i.p. tumor masses reduced by 94 % in the 2 remaining mice, P = 0.00005). Interestingly, when tested against late-stage STO tumors (i.e., 11 days after i.p. DMPM cell injection in mice), CpG-ODN1826 was still able to reduce the growth of i.p. tumor masses by 66 % (P = 0.0009). Peritoneal washings of tumor-bearing mice revealed a strong increase of macrophage infiltration together with a decrease in the presence of B-1 cells and a reduced IgM concentration after CpG-ODN1826 treatment. CONCLUSIONS: Our results indicate that locally administered CpG-ODN1826 is able to markedly affect the growth of both early- and late-stage DMPM orthotopic xenografts in the absence of severe side effects, and suggest a possible clinical role for the agent in the therapy of DMPM.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Mesotelioma/tratamiento farmacológico , Oligodesoxirribonucleótidos/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Cromatografía de Afinidad , Femenino , Citometría de Flujo , Humanos , Inmunidad Humoral/efectos de los fármacos , Inyecciones Intraperitoneales , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Mesotelioma/inmunología , Mesotelioma/patología , Mesotelioma Maligno , Ratones SCID , Estadificación de Neoplasias , Oligodesoxirribonucleótidos/farmacología , Resultado del Tratamiento
4.
Int J Cancer ; 136(2): 299-309, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24866585

RESUMEN

Because available treatments have limited efficacy in triple-negative breast cancer (TNBC), the identification of new therapeutic strategies to improve patients' outcome is urgently needed. In our study, we investigated the effects of the administration of the small molecule selective survivin suppressant YM155, alone or in association with CD34+ cells transduced with a replication-deficient adenovirus encoding the human tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) gene (CD34-TRAIL+ cells), in three TNBC cell models. YM155 exposure significantly impaired TNBC cell growth and selectively modulated survivin expression at both mRNA and protein level. In addition, co-culturing YM155-treated TNBC cells with CD34-TRAIL+ cells resulted in markedly increased cytotoxic effect and apoptotic response in comparison with single treatments. Such a chemosensitizing effect was observed only in TNBC cells inherently expressing DR5 and relied on the ability of YM155 to upregulate DR5 expression through a p38 MAPK- and CHOP-dependent mechanism. YM155/CD34-TRAIL+ combination also showed a significant inhibitory effect on the growth of DR5-expressing TNBC cells following xenotransplantation into NOD/SCID mice, in the absence of toxicity. Overall, our data (i) provide, for the first time, evidence that YM155 sensitizes TNBC cells to CD34-TRAIL+ cells-induced apoptosis by a mechanism involving the downregulation of survivin and the simultaneous p38 MAPK- and CHOP-mediated upregulation of DR5, and (ii) suggest the combination of YM155 with TRAIL-armed CD34+ progenitor cells as a promising therapeutic option for patients with TNBC expressing DR5.


Asunto(s)
Imidazoles/farmacología , Naftoquinonas/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Factor de Transcripción CHOP/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Femenino , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Especies Reactivas de Oxígeno/metabolismo , Survivin , Neoplasias de la Mama Triple Negativas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Chemistry ; 21(39): 13598-608, 2015 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-26286886

RESUMEN

Hsp90 is a molecular chaperone of pivotal importance for multiple cell pathways. ATP-regulated internal dynamics are critical for its function and current pharmacological approaches block the chaperone with ATP-competitive inhibitors. Herein, a general approach to perturb Hsp90 through design of new allosteric ligands aimed at modulating its functional dynamics is proposed. Based on the characterization of a first set of 2-phenylbenzofurans showing stimulatory effects on Hsp90 ATPase and conformational dynamics, new ligands were developed that activate Hsp90 by targeting an allosteric site, located 65 Šfrom the active site. Specifically, analysis of protein responses to first-generation activators was exploited to guide the design of novel derivatives with improved ability to stimulate ATP hydrolysis. The molecules' effects on Hsp90 enzymatic, conformational, co-chaperone and client-binding properties were characterized through biochemical, biophysical and cellular approaches. These designed probes act as allosteric activators of the chaperone and affect the viability of cancer cell lines for which proper functioning of Hsp90 is necessary.


Asunto(s)
Adenosina Trifosfatasas/química , Adenosina Trifosfato/química , Benzofuranos/química , Chaperoninas/química , Proteínas HSP90 de Choque Térmico/química , Adenosina Trifosfatasas/metabolismo , Sitio Alostérico , Fenómenos Bioquímicos , Línea Celular Tumoral , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Hidrólisis , Ligandos , Unión Proteica , Conformación Proteica
6.
J Cell Mol Med ; 17(1): 103-15, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23205553

RESUMEN

The pathogenesis of age-related macular degeneration (AMD) involves demise of the retinal pigment epithelium and death of photoreceptors. In this article, we investigated the response of human adult retinal pigmented epithelial (ARPE-19) cells to 5-(N,N-hexamethylene)amiloride (HMA), an inhibitor of Na(+) /H(+) exchangers. We observed that ARPE-19 cells treated with HMA are unable to activate 'classical' apoptosis but they succeed to activate autophagy. In the first 2 hrs of HMA exposure, autophagy is efficient in protecting cells from death. Thereafter, autophagy is impaired, as indicated by p62 accumulation, and this protective mechanism becomes the executioner of cell death. This switch in autophagy property as a function of time for a single stimulus is here shown for the first time. The activation of autophagy was observed, at a lesser extent, with etoposide, suggesting that this event might be a general response of ARPE cells to stress and the most important pathway involved in cell resistance to adverse conditions and toxic stimuli.


Asunto(s)
Células Epiteliales/metabolismo , Estrés Oxidativo , Epitelio Pigmentado de la Retina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Amilorida/análogos & derivados , Amilorida/farmacología , Apoptosis , Autofagia , Transporte Biológico Activo , Caspasas/genética , Caspasas/metabolismo , Recuento de Células , Línea Celular , Supervivencia Celular , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Etopósido/farmacología , Expresión Génica , Humanos , Especies Reactivas de Oxígeno/agonistas , Especies Reactivas de Oxígeno/metabolismo , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Proteína Sequestosoma-1 , Transducción de Señal , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/metabolismo , Inhibidores de Topoisomerasa/farmacología
7.
Cancer Cell ; 7(5): 457-68, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15894266

RESUMEN

Anticancer agents that selectively kill tumor cells and spare normal tissues are urgently needed. Here, we engineered a cell-permeable peptidomimetic, shepherdin, modeled on the binding interface between the molecular chaperone Hsp90 and the antiapoptotic and mitotic regulator, survivin. Shepherdin makes extensive contacts with the ATP pocket of Hsp90, destabilizes its client proteins, and induces massive death of tumor cells by apoptotic and nonapoptotic mechanisms. Conversely, shepherdin does not reduce the viability of normal cells, and does not affect colony formation of purified hematopoietic progenitors. Systemic administration of shepherdin in vivo is well tolerated, and inhibits human tumor growth in mice without toxicity. Shepherdin could provide a potent and selective anticancer agent in humans.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Diseño de Fármacos , Fragmentos de Péptidos/farmacología , Adenosina Trifosfato/metabolismo , Animales , Proteína con Homeodominio Antennapedia , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Benzoquinonas , Sitios de Unión/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citocromos c/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Fibroblastos/efectos de los fármacos , Productos del Gen tat/genética , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Células HeLa , Proteínas de Homeodominio/genética , Humanos , Proteínas Inhibidoras de la Apoptosis , Lactamas Macrocíclicas , Masculino , Ratones , Ratones SCID , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Moleculares , Imitación Molecular , Proteínas de Neoplasias , Proteínas Nucleares/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Conformación Proteica , Proteínas Proto-Oncogénicas c-bcl-2/genética , Rifabutina/análogos & derivados , Rifabutina/farmacología , Células Madre/efectos de los fármacos , Survivin , Telomerasa/metabolismo , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Lancet Oncol ; 13(8): 810-6, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22819172

RESUMEN

BACKGROUND: The development of new drugs for patients with refractory urothelial cancer is still an unmet medical need. Preclinical evidence lends support to a rationale for targeting of the VEGF or platelet-derived growth-factor axis. We therefore investigated the activity and safety of pazopanib, a multitarget drug with antiangiogenic activity, in patients with urothelial cancer. METHODS: In an open-label, single-group, phase 2 study, patients (aged ≥18 years) with relapsed or refractory urothelial cancer were given pazopanib 800 mg per day, orally. They were treated until disease progression or prohibitive toxicity occurred. The primary endpoint was the proportion of patients who achieved a confirmed objective response, defined as complete or partial response, after independent review, and was analysed by intention to treat. The trial is registered with ClinicalTrials.gov, number NCT01031875. FINDINGS: The trial has been completed. 21 (51%) of 41 patients enrolled were given pazopanib as third-line or further-line treatment. 26 (63%) patients had an Eastern Cooperative Oncology Group performance status of 1 or 2. Seven patients had a confirmed objective response (17·1%, 95% CI 7·2-32·1), all of which were partial responses. The most frequent treatment-related grade 3 adverse events were hypertension (three [7%]), fatigue (two [5%]), and gastrointestinal and vaginal fistulisations (two each [5%]). One patient died as a result of duodenal fistulisation that was related to tissue response of bulky tumour masses. INTERPRETATION: Pazopanib has single-agent activity in patients with heavily pretreated metastatic urothelial cancer, and warrants further study in this setting. Particular attention should be paid to patients with bulky tumour masses adjacent to viscera because fistulisation is probably related to the response to pazopanib and is the most frequent serious adverse event. FUNDING: Fondazione IRCCS Istituto Nazionale dei Tumori provided the grant. GlaxoSmithKline provided the study drug and provided funding for the independent radiological review.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos , Pirimidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Neoplasias Urológicas/tratamiento farmacológico , Urotelio/efectos de los fármacos , Administración Oral , Anciano , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/efectos adversos , Supervivencia sin Enfermedad , Femenino , Humanos , Indazoles , Italia , Estimación de Kaplan-Meier , Modelos Lineales , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Modelos de Riesgos Proporcionales , Pirimidinas/administración & dosificación , Pirimidinas/efectos adversos , Medición de Riesgo , Factores de Riesgo , Sulfonamidas/administración & dosificación , Sulfonamidas/efectos adversos , Factores de Tiempo , Resultado del Tratamiento , Carga Tumoral , Neoplasias Urológicas/mortalidad , Neoplasias Urológicas/patología , Urotelio/patología
9.
Pharmaceuticals (Basel) ; 14(6)2021 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-34072442

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive disease with limited therapeutic options. Here, we pursued a combinatorial therapeutic approach to enhance the activity of selinexor, the first-in-class XPO1 inhibitor, by miR-34a ectopic expression in human TNBC experimental models. Anti-proliferative activity induced by selinexor and miR-34a expression, singly and in combination, was evaluated by MTS assay and cell counting. The effect of treatments on survivin and apoptosis-related proteins was assessed by western blotting and ELISA. The antitumor and toxic effects of individual and combined treatments were evaluated on TNBC orthotopic xenografts in SCID mice. Selinexor consistently showed anti-proliferative activity, although to a variable extent, in the different TNBC cell lines and caused the impairment of survivin expression and intracellular distribution, accompanied by apoptosis induction. Consistent with in vitro data, the XPO1 inhibitor variably affected the growth of TNBC orthotopic xenografts. miR-34a cooperated with selinexor to reduce survivin expression and improved its anti-proliferative activity in TNBC cells. Most importantly, miR-34a expression markedly enhanced selinexor antitumor activity in the less sensitive TNBC xenograft model, in absence of toxicity. Our data form a solid foundation for promoting the use of a miR-34a-based approach to improve the therapeutic efficacy of selinexor in TNBC patients.

10.
Mol Cancer ; 9: 12, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-20092645

RESUMEN

BACKGROUND: Aberrant expression of microRNAs, small non-coding RNA molecules that post-transcriptionally repress gene expression, seems to be causatively linked to the pathogenesis of cancer. In this context, miR-21 was found to be overexpressed in different human cancers (e.g. glioblastoma, breast cancer). In addition, it is thought to be endowed with oncogenic properties due to its ability to negatively modulate the expression of tumor-suppressor genes (e.g. PTEN) and to cause the reversion of malignant phenotype when knocked- down in several tumor models. On the basis of these findings, miR-21 has been proposed as a widely exploitable cancer-related target. However, scanty information is available concerning the relevance of miR-21 for prostate cancer. In the present study, we investigated the role of miR-21 and its potential as a therapeutic target in two prostate cancer cell lines, characterized by different miR-21 expression levels and PTEN gene status. RESULTS: We provide evidence that miR-21 knockdown in prostate cancer cells is not sufficient per se i) to affect the proliferative and invasive potential or the chemo- and radiosensitivity profiles or ii) to modulate the expression of the tumor-suppressors PTEN and Pdcd4, which in other tumor types were found to be regulated by miR-21. We also show that miR-21 is not differently expressed in carcinomas and matched normal tissues obtained from 36 untreated prostate cancer patients subjected to radical prostatectomy. CONCLUSIONS: Overall, our data suggest that miR-21 is not a central player in the onset of prostate cancer and that its single hitting is not a valuable therapeutic strategy in the disease. This supports the notion that the oncogenic properties of miR-21 could be cell and tissue dependent and that the potential role of a given miRNA as a therapeutic target should be contextualized with respect to the disease.


Asunto(s)
MicroARNs/genética , Neoplasias de la Próstata/genética , Línea Celular Tumoral , Cisplatino/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Rayos gamma , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Técnicas de Silenciamiento del Gen , Humanos , Masculino , MicroARNs/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Paclitaxel/farmacología , Próstata/efectos de los fármacos , Próstata/metabolismo , Próstata/patología , Próstata/efectos de la radiación , Neoplasias de la Próstata/patología , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/efectos de la radiación
11.
Cells ; 9(10)2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33023194

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive disease with poor prognosis and limited therapeutic options. Recent advances in the immunotherapy field have enabled the development of new treatment strategies, among which the use of bispecific antibodies (BsAbs), able to redirect T cells against tumors, has shown promising results. In particular, a BsAb that uses TNF-related apoptosis-inducing ligand receptor 2 (TRAIL-R2) as a target was constructed and demonstrated good results in redirecting CD3+ T cells to kill TRAIL-R2-expressing TNBC cells. In the present study, we investigated whether treatment with selinexor, a selective inhibitor of nuclear export (SINE) targeting exportin-1/chromosome maintenance protein 1 (XPO1/CRM1), could potentiate the antitumor activity of this BsAb. In combination experiments, we found that selinexor-exposed TNBC cells exhibited greater growth inhibition when treated with the TRAIL-R2xCD3 BsAb than that expected by simple additivity. Similarly, the apoptosis rate in selinexor/TRAIL-R2xCD3 BsAb-treated TNBC cells was significantly higher than that observed after exposure to either single agent. Together, our results suggest that the combination of selinexor and TRAIL-R2xCD3 BsAb can be a viable anticancer strategy and indicate this treatment as a promising therapeutic option for TNBC patients.


Asunto(s)
Anticuerpos Biespecíficos/fisiología , Hidrazinas/uso terapéutico , Triazoles/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Línea Celular Tumoral , Femenino , Humanos , Hidrazinas/farmacología , Triazoles/farmacología
12.
Methods Mol Biol ; 487: 303-30, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19301654

RESUMEN

The discovery of new cancer-related therapeutic targets is mainly based on the identification of genes involved in pathways selectively exploited in cancer cells, including those leading to unlimited replicative potential, evasion of apoptosis, angiogenesis, tissue invasion and metastatic spread. Potentially, a gene--or a gene product--is recognized as a cancer target whether its modulation in experimental models can specifically modify or revert the cancer phenotype. As soon as RNA interference (RNAi)--a natural gene silencing mechanism--was demonstrated in mammalian cells, it rapidly became an essential means for gene knockdown in preclinical models, making it possible to define the role of several human genes and to identify those specifically involved in the onset and progression of cancer. Owing to its powerful gene-silencing properties, RNAi has been proposed as a useful tool to validate new therapeutic targets and to develop innovative anticancer therapies. This chapter summarizes the findings from recent studies relying on the use of RNAi-based approaches to functionally validate therapeutic targets related to two tumor hallmarks: the unlimited replicative potential (i.e., activation of telomere maintenance mechanisms) and evasion of apoptosis (i.e., up-regulation of anti-apoptotic factors).


Asunto(s)
Apoptosis/fisiología , Neoplasias/terapia , Interferencia de ARN , ARN Interferente Pequeño/uso terapéutico , Telómero/fisiología , Humanos , Neoplasias/genética
13.
Eur J Med Chem ; 168: 253-262, 2019 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-30822713

RESUMEN

Estrogens play a key role in cellular proliferation of estrogen-receptor-positive (ER+) breast cancers (BCs). Suppression of estrogen production by competitive inhibitors of the enzyme aromatase (AIs) is currently one of the most effective therapies against ER + BC. Yet, the development of acquired resistance, after prolonged treatments with AIs, represents a clinical major concern. Serendipitous findings indicate that aromatase may be non-competitively inhibited by clinically employed drugs and/or industrial chemicals. Here, by performing in silico screening on two putative allosteric sites, molecular dynamics and free energy simulations, supported by enzymatic and cell-based assays, we identified five leads inhibiting the enzyme via a non-active site-directed mechanism. This study provides new compelling evidences for the existence of an allosteric regulation of aromatase and for the possibility of exploiting it to modulate estrogens biosynthesis. Such modulation can aptly reduce side effects caused by the complete estrogen deprivation therapy, and, possibly, delay/avoid the onset of resistance.


Asunto(s)
Antineoplásicos/farmacología , Aromatasa/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Regulación Alostérica/efectos de los fármacos , Antineoplásicos/síntesis química , Antineoplásicos/química , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Simulación de Dinámica Molecular , Estructura Molecular , Relación Estructura-Actividad
14.
Sci Rep ; 8(1): 649, 2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29330437

RESUMEN

Somatic mutations of the Estrogen Receptor α (ERα) occur with an up to 40% incidence in ER sensitive breast cancer (BC) patients undergoing prolonged endocrine treatments. These polymorphisms are implicated in acquired resistance, disease relapse, and increased mortality rates, hence representing a current major clinical challenge. Here, multi-microseconds (12.5 µs) molecular dynamics simulations revealed that recurrent ERα polymorphisms (i. e. L536Q, Y537S, Y537N, D538G) (mERα) are constitutively active in their apo form and that they prompt the selection of an agonist (active)-like conformation even upon antagonists binding. Interestingly, our simulations rationalize, for the first time, the efficacy profile of (pre)clinically used Selective Estrogen Receptor Modulators/Downregulators (SERMs/SERDs) against these variants, enlightening, at atomistic level of detail, the key common structural traits needed by drugs able to effectively fight refractory BC types. This knowledge represents a key advancement for mechanism-based therapeutics targeting resistant ERα isoforms, potentially allowing the community to move a step closer to 'precision medicine' calibrated on patients' genetic profiles and disease progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Antagonistas del Receptor de Estrógeno/química , Receptor alfa de Estrógeno/química , Receptor alfa de Estrógeno/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Antagonistas del Receptor de Estrógeno/farmacología , Receptor alfa de Estrógeno/antagonistas & inhibidores , Femenino , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Estructura Secundaria de Proteína
15.
Urol Oncol ; 36(1): 8.e1-8.e8, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28911922

RESUMEN

BACKGROUND: Outcomes of neoadjuvant chemotherapy in patients with muscle-invasive urothelial bladder carcinoma (MIUBC) should be improved. Sorafenib was combined with gemcitabine and cisplatin chemotherapy (SGC) in an open-label, single-arm, phase 2 trial (NCT01222676). PATIENTS AND METHODS: After transurethral resection of the bladder, T2-T4a N0 patients received four cycles of SGC followed by cystectomy. Sorafenib 400mg q12h daily, continuously, was added to standard GC chemotherapy. In a Simon's 2-stage design, the primary endpoint was the pathologic complete response (pT0), assuming H0: ≤0.20 and H1: ≥0.40, with a type I and type II error of 5% and 10%, respectively. RESULTS: From April 2011 to June 2016, 46 patients were enrolled. Pathologic T0 response was obtained in 20 patients (43.5%, 95% CI: 28.9-58.9); pT ≤ 1 in 25 (54.3%, 95% CI: 39.0-69.1). After a median follow-up of 35 months, the median progression-free survival was not reached (NR, interquartile range: 23.6-NR), nor was median overall survival (interquartile range: 30.3-NR). Hematologic and extrahematologic grade 3 to 4 adverse events occurred in 45.6% and 26.1% of patients, respectively. In 29 samples from responders (pT ≤ 1) and nonresponders, different distribution of missense mutations involved DNA-repair genes, RAS-RAF pathway genes, chromatin-remodeling genes, and HER-family genes. ERCC1 immunohistochemical expression was associated with pT ≤ 1 response (P = 0.047). The absence of a comparator arm prevented us to quantify sorafenib contribution. CONCLUSIONS: SGC combination was active in MIUBC, and the identified molecular features included alterations that may help personalize treatment in MIUBC with new more potent targeted agents, combined with chemotherapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Cisplatino/uso terapéutico , Cistectomía/métodos , Desoxicitidina/análogos & derivados , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Cisplatino/administración & dosificación , Cisplatino/farmacología , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Niacinamida/administración & dosificación , Niacinamida/farmacología , Niacinamida/uso terapéutico , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/farmacología , Sorafenib , Resultado del Tratamiento , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/cirugía , Gemcitabina
16.
Dis Markers ; 2018: 9128128, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29682101

RESUMEN

A recent "hot topic" in prostate cancer radiotherapy is the observed association between acute/late rectal toxicity and the presence of abdominal surgery before radiotherapy. The exact mechanism is unclear. Our working hypothesis was that a previous surgery may influence plasma level of inflammatory molecules and this might result in enhanced radiosensitivity. We here present results on the feasibility of monitoring the expression of inflammatory molecules during radiotherapy. Plasma levels of a panel of soluble mediators associated with the inflammatory response were measured in prostate cancer patients undergoing radical radiotherapy. We measured 3 cytokines (IL-1b, IL-6, and TNF alpha), 2 chemokines (CCL2 and CXCL8), and the long pentraxin PTX3. 20 patients were enrolled in this feasibility evaluation. All patients were treated with IMRT at 78 Gy. 3/20 patients reported grade 2 acute rectal toxicity, while 4/20 were scored as grade 2 late toxicity. CCL2 was the most interesting marker showing significant increase during and after radiotherapy. CCL2 levels at radiotherapy end could be modelled using linear regression including basal CCL2, age, surgery, hypertension, and use of anticoagulants. The 4 patients with late toxicity had CCL2 values at radiotherapy end above the median value. This trial is registered with ISRCTN64979094.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias de la Próstata/sangre , Traumatismos por Radiación/sangre , Radioterapia de Intensidad Modulada/efectos adversos , Anciano , Proteína C-Reactiva/metabolismo , Quimiocina CCL2/sangre , Humanos , Interleucina-1/sangre , Interleucina-6/sangre , Interleucina-8/sangre , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata/radioterapia , Traumatismos por Radiación/etiología , Componente Amiloide P Sérico/metabolismo , Factor de Necrosis Tumoral alfa/sangre
17.
Biochem Pharmacol ; 73(6): 773-81, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17188248

RESUMEN

Based on previous evidence indicating a selective cytotoxic activity of the mixed phosphine gold complex chlorotriphenylphosphine-1,3-bis(diphenylphosphino)propanegold(I) for melanoma cells, we investigated the cellular bases of its antiproliferative effect in a panel of human melanoma cell lines (JR8, SK-Mel-5, Mel-501, 2/60, 2/21 and GRIG). The drug consistently induced a dose-dependent inhibition of cell growth, with IC50 values ranging from 0.8 to 2.3 microM and, when tested under the same experimental conditions, its cytotoxic activity was higher than (from 2- to 5-fold) or comparable to that of cisplatin as a function of cell lines. The ability of the gold complex to activate programmed cell death was assessed in JR8 and 2/60 cells, and a dose-dependent increase in cells with an apoptotic nuclear morphology was observed in both cell lines (up to 40 and 66% of the overall cell population, for JR8 and 2/60 cell lines, respectively). Such an apoptotic response was mediated by a dose-dependent loss of mitochondrial membrane potential, cytochrome c and Smac/DIABLO release from mitochondria into cytosol and enhanced caspase-9 and caspase-3 catalytic activity. A reduced or completely abrogated expression of the anti-apoptotic proteins c-IAP1, XIAP and survivin in drug-treated cells was also observed. Overall, results from the study indicate that chlorotriphenylphosphine-1,3-bis(diphenylphosphino)propanegold(I) markedly inhibits melanoma cell growth by inducing mitochondria-mediated apoptosis and suggest it as a good candidate for additional evaluation as an anticancer agent against melanoma.


Asunto(s)
Antineoplásicos/farmacología , Melanoma/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Compuestos Orgánicos de Oro/farmacología , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Melanoma/patología
18.
Cell Oncol ; 29(6): 453-66, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18032822

RESUMEN

BACKGROUND: The biology of malignant peritoneal mesothelioma (MPM) is largely unknown. In the present study, we assessed the expression of survivin and other members of the inhibitors of apoptosis proteins (IAP) family (IAP-1, IAP-2 and X-IAP) in a series of 32 MPM surgical specimens and investigated the effects of survivin knockdown in an established MPM cell line. METHODS: Expression of different IAPs was measured by immunohistochemistry. MPM cells were transfected with a small-interfering RNA (siRNA) targeting survivin mRNA and analyzed for survivin expression, growth rate, and ability to undergo spontaneous and drug (cisplatin, doxorubicin)-induced apoptosis. RESULTS: Survivin expression was observed in 29 (91%) surgical MPM specimens, whereas the positivity rate for the other IAPs ranged from 69% to 100%. Transfection of MPM cells with the survivin siRNA induced a marked inhibition of survivin protein expression, a time-dependent decline in cell growth and an enhanced rate of spontaneous and drug-induced apoptosis, with a concomitant increase in the catalytic activity of caspase-9. CONCLUSION: Our results show for the first time that survivin, as well as other IAPs, is largely expressed in clinical MPMs and suggest that strategies aimed at down-regulating survivin may provide a novel approach for the treatment of the malignancy.


Asunto(s)
Mesotelioma/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/fisiología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Neoplasias Peritoneales/genética , Adulto , Anciano , Proteínas Reguladoras de la Apoptosis/metabolismo , Caspasa 9/metabolismo , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/fisiología , Masculino , Mesotelioma/química , Persona de Mediana Edad , Neoplasias Peritoneales/química , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Survivin
19.
Methods Mol Biol ; 361: 239-63, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17172716

RESUMEN

In recent years expanding knowledge about basic biology and a detailed understanding of the molecular pathways involved in tumor cell growth and progression have allowed the identification of numerous genes as potential therapeutic targets. Studies in which the expression of these genes was manipulated by antisense strategies have provided clues as to how we can intervene to specifically kill tumor cells or sensitize them to conventional chemical and physical antitumor therapies. Such tumor specificity can only be obtained by exploiting a basic difference between normal and malignant cells. In this context, targeting cytoprotective factors such as telomerase and survivin is particularly attractive because of their almost selective expression in tumor cells and their proven association with disease progression. This chapter summarizes the results obtained with ribozymes and small-interfering RNAs in the functional validation of these two targets in cell cultures and animal tumor models.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , ARN Catalítico/farmacología , ARN Interferente Pequeño/farmacología , Telomerasa/antagonistas & inhibidores , Animales , Antineoplásicos/metabolismo , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Proteínas Inhibidoras de la Apoptosis , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/metabolismo , Interferencia de ARN/efectos de los fármacos , ARN Catalítico/genética , ARN Catalítico/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Survivin , Telomerasa/genética , Telomerasa/metabolismo
20.
Mol Cancer Ther ; 5(1): 179-86, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16432177

RESUMEN

Survivin is an antiapoptotic gene, which is overexpressed in most human tumors and involved in mitotic checkpoint control. Recent evidence points to an essential role for heat shock protein 90 (Hsp90) in survivin function regulation. Although the survivin-Hsp90 association may promote tumor cell proliferation, it may also suggest new opportunities for the design of novel anticancer approaches. We evaluated the effect of small interfering RNA (siRNA)-mediated inhibition of survivin on the proliferative potential of prostate cancer cells and their sensitivity to the Hsp90 inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG). Human androgen-independent prostate cancer cell lines (DU145 and PC-3) were transfected with four 21-mer double-stranded siRNAs (100 nmol/L) directed against different portions of survivin mRNA. After transfection, cells were collected and analyzed for survivin mRNA and protein expression, cell proliferation rate, ability to undergo apoptosis, and sensitivity to 17-AAG. Transfection of prostate cancer cells with siRNAs induced a variable extent of inhibition of survivin mRNA expression (39-60% compared with controls), which was paralleled by a 38% to 75% reduction in survivin protein abundance. The three siRNAs able to induce the greatest inhibition of survivin expression also significantly reduced cell proliferation and enhanced the rate of apoptosis, with a concomitant increase in caspase-9 activity. Sequential treatment with siRNA and 17-AAG induced supra-additive antiproliferative effects in all cell lines, with an enhanced caspase-9-dependent apoptotic response. These findings suggest that combined strategies aimed at interfering with the survivin-Hsp90 connection may provide novel approaches for treatment of androgen-independent prostate cancer.


Asunto(s)
Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , ARN Interferente Pequeño/farmacología , Rifabutina/análogos & derivados , Apoptosis/efectos de los fármacos , Apoptosis/genética , Benzoquinonas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Proteínas Inhibidoras de la Apoptosis , Lactamas Macrocíclicas , Masculino , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , Proteínas de Neoplasias/efectos de los fármacos , Neoplasias de la Próstata/patología , ARN Interferente Pequeño/genética , Rifabutina/farmacología , Survivin , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA